Back to Journals » International Journal of Chronic Obstructive Pulmonary Disease » Volume 19

Causal Involvement of Immune Cells in Chronic Obstructive Pulmonary Disease: A Mendelian Randomization Study

Authors Guan T , Qin Y, Qu N, Pan Y

Received 11 March 2024

Accepted for publication 29 June 2024

Published 9 July 2024 Volume 2024:19 Pages 1603—1611

DOI https://doi.org/10.2147/COPD.S460342

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Dr Jill Ohar



Tiefa Guan,1,2,* Yibing Qin,3,* Nini Qu,3 Yushuo Pan1

1First Clinical College, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning Province, People’s Republic of China; 2Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning Province, People’s Republic of China; 3Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning Province, People’s Republic of China

*These authors contributed equally to this work

Correspondence: Nini Qu, Email [email protected]

Background: The immune cells play a substantial role in the development and progression of chronic obstructive pulmonary disease (COPD). We aim to investigate the causal involvement of immune cells in COPD via a Mendelian randomization (MR) analysis.
Methods: Published genome-wide association studies (GWAS) statistics on immune cells were analyzed, with genetic variants identified as instrumental variables (IVs). Inverse-variance weighting (IVW), weighted median, and MR-Egger regression methods were employed, along with simple mode and weighted mode adopted in the two-sample MR analysis. Sensitivity analysis was conducted to examine the heterogeneity, horizontal pleiotropy, and stability of the causal relationship.
Results: IVW results suggested that CCR2 on CD62L+ plasmacytoid dendritic cells (DC), CCR2 on plasmacytoid DC, CD11b on CD66b++ myeloid cells, CD19 on CD20− CD38− CD24+ memory B cell subset, CD25 on transitional B cells, and CD25++CD8br %CD8br T cells were risk factors for the development of COPD. Besides, CD127 on effector memory-like cytotoxic T lymphocytes lacking expression of co-stimulatory molecule 28 (CD28-EM CTLs) and HLA DR+ NK ACs expressing human leukocyte antigen DR molecules while being natural killer cells (%NK ACs) were protective factors for COPD.
Conclusion: This study unveiled a causal relationship between immune cell phenotype and COPD. These findings offer new insights for the prevention and treatment of COPD using COPD-associated immune cells.

Keywords: chronic obstructive pulmonary disease, immune cells, causal inference, Mendelian randomization

Introduction

Chronic obstructive pulmonary disease (COPD) is a leading cause of death worldwide characterized by progressive airflow restriction and accelerated lung function decline.1 A long-term course of COPD eventually damages lung epithelial cells and lung tissue, leading to airway inflammation, increased mucus secretion, peribronchial fibrosis, and airway remodeling.2 Generally, chronic innate and adaptive inflammatory immune responses are triggered by COPD.3 COPD-induced inflammatory responses involve various components of innate immunity, such as eosinophils, neutrophils, macrophages, mast cells, natural killer (NK) cells, and dendritic cells (DC), as well as components of adaptive immunity like T lymphocytes and B lymphocytes.4 Although the importance of immune cells in the pathology of COPD has been established, its causal relationship with COPD remains largely unclear. This study aims to investigate the causal involvement of immune cell phenotypes in the development of COPD by a Mendelian randomization (MR) analysis, thus providing novel therapeutic targets for COPD.

Materials and Methods

Data Sources

Genetic signatures in immune cell traits, involving 22 million variants and their impact on 731 immune cell phenotypes in a cohort of 3757 individuals of Sardinian descent were available from a published literature.5 Additionally, summary data on 13,530 COPD patients and 454,945 controls of European ancestry, with 24,180,654 single nucleotide polymorphisms (SNPs) were obtained from the IEU OpenGWAS project (https://gwas.mrcieu.ac.uk/, accessed on November 5, 2023). An ethical approval of the present study was not necessary because of the publicly available datasets.

Selection of Tool Variables

Optimal instrumental variables (IVs) were selected following the rigorous screening criteria, aiming to ensure the authenticity and accuracy of our findings. Briefly, summary-level genome-wide association study (GWAS) statistics on immune cells were screened, and SNPs with a significance threshold of P<5×10−5 were selected. Following the independence assumption of MR, a coefficient R2 of 0.001 and clump distance of 10,000 kb maximized their efforts to avoid any potential linkage disequilibrium IVs. Further, palindromic SNPs were excluded to guarantee consistent alleles for each SNP in both the immune cell group and COPD group. The process of selecting the optimal IVs was depicted in (Figure 1).

Figure 1 Flowchart of MR analysis.

MR Assumptions

A valid MR analysis required that IVs should adhere to the following three fundamental assumptions: (1) Independence assumption: the selected IVs were independent of confounding factors influencing immune cells and COPD; (2) Association assumption: IVs were robustly associated with COPD; (3) Exclusivity assumption: IVs were associated with COPD through immune cells only (Figure 2).

Figure 2 Principal assumptions of MR.

Statistical Processing

Two-sample MR and MR-PRESSO in R 4.320 were utilized for the MR analysis and sensitivity analysis, respectively. The F statistic was employed to assess the strength of correlation between IVs and COPD, where an F value of greater than 10 indicated a satisfaction of the association assumption.

MR Analysis

Inverse-variance weighting (IVW), MR-Egger regression, weighted median, simple model, and weighted model were employed in the present MR analysis of inferring a causal relationship between immune cells and COPD. In the absence of horizontal pleiotropy, IVW method approached a weighed mean of the reciprocal of outcome variance, rather than the consideration of an intercept term in the regression analysis. IVW results were considered as the primary outcomes. Depending on heterogeneity presence or absence, either a fixed-effects or a random-effects model was adopted in IVW. MR-Egger regression provided consistent estimates even when all IVs exhibited gene pleiotropy. The weighted median was excellent in consistently estimating causal relationships even when more than 50% of IVs were invalid. The simple model offered a robustness against genetic pleiotropy,6 and the weighted model was featured by the high sensitivity to challenge bandwidth selection during model estimation.7 Therefore, IVW, MR-Egger regression and weighted median method were used in the MR analysis, with the simple and weighted models adopted as supplementary methods. A significance level was determined at p<0.05.

Sensitivity Analysis

Heterogeneity was assessed using Cochran’s Q test. The MR-PRESSO was employed to identify and remove outliers, followed by reanalysis. MR-Egger analysis allowed the evaluation of horizontal pleiotropy by testing the intercept. The robustness of MR results was evaluated using the leave-one-out approach. A significance level was determined at p<0.05.

Results

Optimal IVs

After screening GWAS statistics on immune cells, a total of 18,621 SNPs with F value of greater than 10 were selected as IVs.

MR Analysis

IVW data revealed that CCR2 on CD62L+ plasmacytoid DC (OR=1.039, 95% CI 1.008, 1.065, P<0.05), CCR2 on plasmacytoid DC (OR=1.036, 95% CI 1.001, 1.003, P<0.05), CD11b on CD66b++ myeloid cells (OR=1.033, 95% CI 1.004, 1.063, P<0.05), CD19 on CD20− CD38− (OR=1.084, 95% CI 1.038, 1.131, P<0.05), CD24 on memory B cells (OR=1.027, 95% CI 1.004, 1.051, P<0.05), CD25++ CD8br %CD8br (OR=1.042, 95% CI 1.002, 1.084, P<0.05), CD27 on CD24+ CD27+ (OR=1.040, 95% CI 1.016, 1.065, P<0.05), CD27 on switched memory cells (OR=1.033, 95% CI 1.006, 1.061, P<0.05), CD27 on unswitched memory cells (OR=1.036, 95% CI 1.007, 1.066, P<0.05), CD28+ CD45RA− CD8br %T cells (OR=1.015, 95% CI 1.000, 1.029, P<0.05), CD33dim HLA DR+ CD11b− %CD33dim HLA DR+ (OR=1.017, 95% CI 1.003, 1.032, P<0.05), CD62L− CD86+ myeloid DC %DC (OR=1.023, 95% CI 1.001, 1.047, P<0.05), CD62L− myeloid DC AC (OR=1.044, 95% CI 1.009, 1.080, P<0.05), CD62L on monocytes (OR=1.028, 95% CI 1.002, 1.055, P<0.05), HLA DR on plasmacytoid DC (OR=1.020, 95% CI 1.003, 1.037, P<0.05), IgD+ CD38br %B cells (OR=1.038, 95% CI 1.007, 1.069, P<0.05), myeloid DC AC (OR=1.038, 95% CI 1.015, 1.061, P<0.05) and NKT %lymphocytes (OR=1.042, 95% CI 1.012, 1.074, P<0.05) were risk factors for COPD. In addition, CD127 on CD28− CD8br (OR=0.957, 95% CI 0.917, 0.998, P<0.05), CD127 on CD28+ CD45RA− CD8br (OR=0.970, 95% CI 0.946, 0.995, P<0.05), CD14+ CD16+ monocyte AC (OR=0.968, 95% CI 0.940, 0.998, P<0.05), CD19 on IgD− CD38br (OR=0.955, 95% CI 0.919, 0.992, P<0.05), CD19 on PB/PC (OR=0.940, 95% CI 0.903, 0.978, P<0.05), CD25hi %T cells (OR=0.965, 95% CI 0.934, 0.997, P<0.05), CD3− lymphocyte AC (OR=0.945, 95% CI 0.906, 0.986, P<0.05), CD3 on CM CD8br (OR=0.957, 95% CI 0.924, 0.996, P<0.05), CD3 on EM CD8br (OR=0.965, 95% CI 0.942, 0.989, P<0.05), CD3 on naive CD8br (OR=0.95, 95% CI 0.994, 0.999, P<0.05), CD4+ CD8dim %leukocytes (OR=0.943, 95% CI 0.894, 0.995, P<0.05), CD4+ CD8dim %lymphocytes (OR=0.943, 95% CI 0.904, 0.984, P<0.05), CD45 on Im myeloid derived suppressor cells (MDSCs) (OR=0.965, 95% CI 0.941, 0.990, P<0.05), CD62L on granulocytes (OR=0.944, 95% CI 0.908, 0.981, P<0.05), CD80 on granulocytes (OR=0.967, 95% CI 0.944, 0.991, P<0.05), HLA DR on monocytes (OR=0.978, 95% CI 0.955, 1.000, P<0.05), HLA DR+ NK AC (OR=0.951, 95% CI 0.916, 0.987, P<0.05), HLA DR+ T cell%T cells (OR=0.977, 95% CI 0.958, 0.996, P<0.05), naive CD8br AC (OR=0.964, 95% CI 0.934, 0.996, P<0.05), SSC−A on HLA DR+ T cells (OR=0.957, 95% CI 0.926, 0.989, P<0.05) and TCRgd %T cells (OR=0.965, 95% CI 0.937, 0.994, P<0.05) served as protective factors for COPD (Figure 3).

Figure 3 A forest map visualizing a causal relationship between immune cells and COPD.

Sensitivity Analysis

A significance level of p<0.05 obtained in the heterogeneity test suggested no significant heterogeneity among the combined IVs, and a fixed-effects model was then employed for all IVW analyses. No outliers and horizontal pleiotropy were detected by MR-PRESSO and MR-Egger, respectively. Additionally, a leave-one-out approach revealed that neither of an individual SNP had a significant impact on the robustness of our findings. Therefore, the MR analysis unveiled a causal involvement of immune cells in the development of COPD.

Discussion

Tissue damage and disease severity of COPD are associated with the formation of tertiary lymphoid organs (TLOs).2,8 Lymphoid follicles (LFs) are the predominant type of TLOs, consisting of a large population of B lymphocytes surrounded by DC and small numbers of CD4+ and CD8+ cells.9 B cells in LFs interact with certain subsets of CD4+ T cells and thus experience phenotypic transformation and maturation. Their interactions further trigger the release of inflammatory factors, serving as an inducer for tissue injury. Therefore, an effective suppression of inducers for inflammatory responses is considered as a crucial step in blocking immune responses in LFs and the onset of COPD.10 Various types of immune cells are involved in the immune reactions that either stimulate or inhibit the pathogenesis of COPD. Using publicly available databases, we investigated the causal relationship between 731 immune cell traits and COPD by an MR analysis. A total of 39 immunophenotypes across 4 immune cell traits of median fluorescence intensity (MFI), relative cell count (RC), absolute cell count (AC) and morphological parameters (MP) that exhibited significant causal effects on COPD.

Seven immune cell phenotypes associated with DC were found as risk factors for the onset of COPD, including CCR2 on CD62L+ plasmacytoid DC, CCR2 on plasmacytoid DC, HLA DR on plasmacytoid DC, CD62L-CD86 + myeloid DC AC, CD62L-CD86 + myeloid DC %DC, CD62L on monocytes, and myeloid DC AC. Two granulocyte phenotypes, namely CD62L on granulocytes and CD80 on granulocytes, were identified as protective factors for COPD. Plasmacytoid (pDC) and myeloid DC (mDC1 and mDC2) are bone-marrow-derived and blood-derived cells, respectively. Both of them can directly or indirectly participate in the process of lung dysfunction and mediation of downstream T lymphocytes11. The activated adaptive immune system is also involved in the pathogenesis of COPD through inducing inflammation responses or antigen presentation.12 Although ranking the highest proportion, mDC2, alongside mDC1 and pDC in the lung tissue are quantitatively similar between COPD patients and healthy volunteers. Expression levels of costimulatory molecules, however, are significant different between COPD patients and healthy controls. Specifically, CD40 on mDC1, CD86 on mDC2, CD40 on mDC2, CD80 on pDC and CD40 on pDC increase with the increasing severity of COPD. Upregulation of costimulatory molecules also result in a stronger stimulation towards downstream T cells.13 It is found that mDC2 exerts an excellent performance in inducing helper T lymphocytes.8,14 The higher absolute values of mDC and pDC in the lung lavage fluid of COPD smokers compared to those of non-smokers are indicative of the importance of tobacco exposure in the pathogenesis of COPD14. After 4 hours of smoking, an increase in alveolar lavage fluid mDC and decrease in blood mDC among smokers suggested that the selective migration of mDC to the airway and its contribution to antigen presentation15. Co-stimulatory molecules CD62L, CD80, and CD86 expressed on the surface of immune cells play a vital role in mediating cell-cell interactions with lymphocytes. Human leukocyte antigen (HLA) serves as a key molecule involved in DC-mediated antigen presentation. CCR2 regulates cell-cell interactions by binding to specific chemokines, and the frequency of CCR2-related gene carriers is positively correlated with the incidence of COPD.16 Overall, DC-induced immune function affects the formation of LFs by mediating downstream immune cell responses, thus influencing the development and progression of COPD.

T cells and B cells are the primary cellular components involved in the adaptive immune response. In our study, most T lymphocyte phenotypes were identified as protective factors for COPD, including CD127 on CD28+ CD45RA− CD8br, CD25hi %T cells, CD28+ CD45RA− CD8br %T cells, CM-CD3 on naive EM-CD8br, naive AC-CD8br, AC-CD3 lymphocytes, leukocyte% of CD4+ and dimly expressed% of lymphocytes in HLA DR+ T cells. However, TCRgd %T cells and highly expressed markers (CD25++ and dimly expressed) were risk factors for COPD. With the increasing severity of COPD, there are increased percentages of small airways containing immune cells (eg, CD4+, CD8+, B lymphocytes) and lymphoid aggregates consisting of follicles macrophages and neutrophils.2 Differentiated from undifferentiated T lymphocytes after antigen presentation by T cell receptors (TCRs), the phenotype of surface marker helper T cells primarily assists B-cells to produce antibodies and enhance humoral immunity, and secondarily supports other lymphocyte functions of inflammatory cell activity through releasing cytokines.17 CD8+ cells represent inhibitory and cytotoxic T lymphocytes that suppress B lymphocyte activity, antibody synthesis and secretion, and T cell proliferation.18 Therefore, an increase in the population of CD8+ cells is not conducive to antibody production or inflammation control, but signifies a subgroup of cells responsible for clearing infected or damaged cells.19 CD4+ T lymphocytes, predominantly the Th1 subtype, accumulate in the airway and lung tissue of COPD patients. They are inducers of inflammatory cytokines and chemokines and ultimately lead to emphysema.20 Besides CD4+ T lymphocytes, CD8+ T lymphocytes (particularly the Tc1 population) also increase in the peripheral blood of COPD patients21. Influenced by proinflammatory cytokines, CD4+CD25−Foxp3+-labeled T cells promote the proliferation and differentiation of naive CD4+ T cells into proinflammatory Th17 effector cells in COPD. These specific T cell phenotypes are responsible for lung inflammation via assisting leukotriene formation.21–23 Regulatory T cells (Tregs) are a specialized subset of CD4+ T lymphocytes expressing CD25, presenting anti-inflammatory and immunomodulatory properties. Upon activation, Tregs inhibit the proliferation of both CD4+ and CD8+T cells through cell-contact dependent mechanisms and the secretion of cytokines (primarily IL-10). The protective function of Tregs in regulating the immune system and inhibiting inflammatory responses, however, is diminished in COPD patients.24 Maturation (CD45RA, CD45R0) and activation (CD28) patterns on both CD4+ and CDS-T lymphocytes are considered as crucial cellular phenotypes involved in the pathogenesis of COPD2. A higher proportion of CD8+CD45RA+ T lymphocytes in the bronchoalveolar lavage fluid (BALF) of COPD patients, that is, a maturation pattern, predicts a greater potential of tissue damage. Down-regulation of CD45RA and up-regulation of CD28 may potentially inhibit the development of COPD,25,26 which were consistent with our identifications of risk and protective factors for COPD. A phenotype analysis of peripheral blood CD4+T cells in patients experiencing acute exacerbation of COPD (AECOPD) revealed that the abnormal activation patterns of Th1, Th17, and Treg cells, and an imbalance between Th1/Th2 cells altogether contribute to a Th1-dominant enhanced immunoinflammatory response.27 Both CD4+ and CD8+T lymphocytes increase in either stable or acute COPD patients. Further investigation, nevertheless, is needed to determine the causal relationship between different phenotypes of immune cells and the progression of COPD.

Our findings demonstrated that CD19 expression on CD20− CD38− cells, CD24 expression on memory B cells, CD25 expression on transitional B cells, CD27 expression on CD24+CD27+ cells, and IgD+CD38br %B cells were risk factors for COPD, while the presence of CD19 on IgD−CD38br cells was a protective factor. Plasma cells (PCs) and memory B cells are two distinct types of lung-resident B cells. The former is capable of producing mainly IgA and IgM antibodies and triggering their secretion into the airway lumen, and the latter generated during the primary lung infection elicits a secondary immune response.28 LFs formed in responsiveness to the activation of B cells reside in B cells and aggregate in small airways, showing a close association with emphysema in patients with chronic COPD.29 CD27+ LFs can also be found within the pulmonary parenchyma of COPD patients30. Characterized by an excessive activation of B cells in the lungs, emphysema eventually Results in the increase in immature, memory, and antibody-producing B cells, and induces a vicious circle to further exacerbates emphysematous pathology.31,32 Overexpression of B-cell stimulating factor (BAFF) in LFs causes lung inflammation exacerbation and alveolar destruction through promoting B cell proliferation. Produced by B cells, IL-10 is a cytokine that adds immune responses in neutrophils and macrophages (fuel) to the chronic airway inflammation (flame).33 IgA produced by B cells impairs mucosal immunity and aggravates the progression of COPD.34 Currently, the relationship between B cell-mediated immune response and the pathogenesis of COPD remains unclear. B-cell-triggered immunoinflammatory response is generally considered as a vital event accelerating the development of COPD. Therefore, inhibiting B cell activation is a promising therapeutic strategy for COPD.

Our findings suggested that the presence of CD14+ CD16+ monocyte antigen-presenting cells (APCs) and high expression of HLA DR on monocytes were protective factors against COPD. Three subsets of myeloid cells reside in the lungs, namely resident alveolar macrophages (AMs), resident interstitial macrophages (IMs), and blood monocytes.35 Lung AMs show typical phenotypes of positive CD14 and CD16.36 They are responsible for clearing alveolar surface active substances, inhibiting adaptive immunity, regulating inflammatory cell recruitment, and generating reactive oxygen species (ROS).37 In COPD patients, AMs exhibit altered secretion patterns of pro-inflammatory mediators like TNF-α, IL-1β, IL-6, and CCL3 upon TLR stimulation.38,39 Both monocytes and AMs are potent producers of matrix metalloproteinases (MMPs) that contribute to elastolysis and destruction of alveolar tissue in the lungs.40 Furthermore, the phenotype of CD14+HLA-DR+ macrophages is the major source of TNF-α. Their detection in sputum samples of COPD patients indicates the involvement of high-level TNF-α in the onset of COPD. Consistently, our findings also highlighted the role of activated AMs in inhibiting the development of COPD.

In the present study, we for the first time unveiled a causal relationship between immune cells and COPD via an MR analysis. Rigorous measures were adopted to exclude SNPs associated with COPD and immune cells from the analysis, minimizing potential confounding effects. In addition, our study provided a relatively large sample size for exposures and results, enhancing its reliability. However, limitations of the study existed. First of all, genetic and immune cell statistics were only from European populations. Our findings may not applicable to other countries and populations. Secondly, the immunophenotype of COPD patients varies with disease severity and stage. Finally, it was a methodology-based analysis, and more extensive longitudinal cohort studies and long-term follow-upobservations are needed to fully evaluate the potential of immune cells in the prevention and treatment of COPD.

In recent years, a dynamic balance between pro-inflammatory and anti-inflammatory signals and the relationship between innate immune cells and adaptive immune cells have been well concerned in the prevention and treatment of COPD. COPD breaks the cross-talk among various immune cells and the balance of pro-inflammatory and anti-inflammatory responses, resulting in chronic airway inflammation, airway remodeling and tissue damage.41 Fibroblasts increase in the lung tissue of COPD patients, and moreover, Th1/Tc1 cells, Th17 cells, activated B cells, and NK cells present within lung fibroblasts induce specific tissue damage and inflammation. Due to the downregulation or loss-of-function of TGF-β1 (transforming growth factor-beta 1), FOXP3 (forkhead box P3), CTLA-4 (cytotoxic T-lymphocyte-associated protein 4) and antioxidants in the lung tissue of COPD patients, an anti-inflammatory response is diminished.28 Although a complex interaction pattern between the immune system and COPD has been established, immune response processes involved with different immune cell phenotypes remain largely unclear. Through an MR analysis, our study identified risk and protective immune cell traits for COPD while minimizing confounding factors, providing new avenues to the prevention and management of COPD.

Data Sharing Statement

The GWAS summary data were acquired from the publicly accessible online platform (https://gwas.mrcieu.ac.uk/). The analyses of the GWAS summary data were conducted using application R version 4.2.1.

Ethical Approval

Summary statistics for the studies used for analysis were composed and obtained from published studies. All studies have received prior approval from their respective institutional review boards (IRBs). The institutional Review Board of The Second Affiliated Hospital of Liaoning University of Traditional Chinese Medicine approved the protocol for this study, and as per their guidelines, this study exclusively utilized publicly available data without using any individual-level data. Therefore, no additional IRB approval was necessary.

Acknowledgment

Tiefa Guan and Yibing Qin are co-first authors for this work. We express our gratitude to the IEU Open GWAS Project for their invaluable efforts in collecting and archiving the GWAS summary statistics related to immune cells.

Funding

The work was supported by the National Natural Science Foundation of China (Grant number 82374378). Seedling project of the Second Affiliated Hospital of Liaoning University of Traditional Chinese Medicine (Grant number 2023-LZYY-1-07).

Disclosure

The authors report no conflicts of interest in this work.

References

1. Eapen MS, Myers S, Walters EH, Sohal SS. Airway inflammation in chronic obstructive pulmonary disease (COPD): a true paradox. Expert Rev Respir Med. 2017;11(10):827–839. doi:10.1080/17476348.2017.1360769

2. Hogg JC, Chu F, Utokaparch S, et al. The nature of small-airway obstruction in chronic obstructive pulmonary disease. N Engl J Med. 2004;350(26):2645–2653. doi:10.1056/NEJMoa032158

3. Hogg JC. Pathophysiology of airflow limitation in chronic obstructive pulmonary disease. Lancet. 2004;364(9435):709–721. doi:10.1016/s0140-6736(04)16900-6

4. Racanelli AC, Kikkers SA, Choi AMK, Cloonan SM. Autophagy and inflammation in chronic respiratory disease. Autophagy. 2018;14(2):221–232. doi:10.1080/15548627.2017.1389823

5. Orrù V, Steri M, Sidore C, et al. Complex genetic signatures in immune cells underlie autoimmunity and inform therapy. Nat Genet. 2020;52(10):1036–1045. doi:10.1038/s41588-020-0684-4

6. Milne RL, Kuchenbaecker KB, Michailidou K, et al. Identification of ten variants associated with risk of estrogen-receptor-negative breast cancer. Nat Genet. 2017;49(12):1767–1778. doi:10.1038/ng.3785

7. Hartwig FP, Davey Smith G, Bowden J. Robust inference in summary data Mendelian randomization via the zero modal pleiotropy assumption. Int J Epidemiol. 2017;46(6):1985–1998. doi:10.1093/ije/dyx102

8. Naessens T, Morias Y, Hamrud E, et al. Human lung conventional dendritic cells orchestrate lymphoid neogenesis during chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2020;202(4):535–548. doi:10.1164/rccm.201906-1123OC

9. Brusselle GG, Demoor T, Bracke KR, Brandsma CA, Timens W. Lymphoid follicles in (very) severe COPD: beneficial or harmful? Eur Respir J. 2009;34(1):219–230. doi:10.1183/09031936.00150208

10. Freeman CM, Curtis JL. It’s complicated: lung dendritic cells in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2020;202(4):479–481. doi:10.1164/rccm.202004-0899ED

11. Freeman CM, Curtis JL. Lung dendritic cells: shaping immune responses throughout chronic obstructive pulmonary disease progression. Am J Respir Cell Mol Biol. 2017;56(2):152–159. doi:10.1165/rcmb.2016-0272TR

12. Van Pottelberge GR, Bracke KR, Van den Broeck S, et al. Plasmacytoid dendritic cells in pulmonary lymphoid follicles of patients with COPD. Eur Respir J. 2010;36(4):781–791. doi:10.1183/09031936.00140409

13. Freeman CM, Martinez FJ, Han MK, et al. Lung dendritic cell expression of maturation molecules increases with worsening chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2009;180(12):1179–1188. doi:10.1164/rccm.200904-0552OC

14. Stoll P, Heinz AS, Bratke K, et al. Impact of smoking on dendritic cell phenotypes in the airway lumen of patients with COPD. Respir Res. 2014;15(1):48. doi:10.1186/1465-9921-15-48

15. Lommatzsch M, Bratke K, Knappe T, et al. Acute effects of tobacco smoke on human airway dendritic cells in vivo. Eur Respir J. 2010;35(5):1130–1136. doi:10.1183/09031936.00090109

16. Bai J, Song H, Cai C, Zhang M, Xu S, Tan J. The association of monocyte chemotactic protein-1 and CC chemokine receptor 2 gene variants with chronic obstructive pulmonary disease. DNA Cell Biol. 2012;31(6):1058–1063. doi:10.1089/dna.2011.1520

17. Lemaire F, Audonnet S, Perotin JM, et al. The elastin peptide VGVAPG increases CD4(+) T-cell IL-4 production in patients with chronic obstructive pulmonary disease. Respir Res. 2021;22(1):14. doi:10.1186/s12931-020-01609-4

18. Zhuang H, Li N, Chen S, et al. Correlation between level of autophagy and frequency of CD8(+) T cells in patients with chronic obstructive pulmonary disease. J Int Med Res. 2020;48(9):300060520952638. doi:10.1177/0300060520952638

19. Williams M, Todd I, Fairclough LC. The role of CD8 + T lymphocytes in chronic obstructive pulmonary disease: a systematic review. Inflamm Res. 2021;70(1):11–18. doi:10.1007/s00011-020-01408-z

20. Albertson TE, Chenoweth JA, Pearson SJ, Murin S. The pharmacological management of asthma-chronic obstructive pulmonary disease overlap syndrome (ACOS). Expert Opin Pharmacother. 2020;21(2):213–231. doi:10.1080/14656566.2019.1701656

21. Xue W, Ma J, Li Y, Xie C. Role of CD(4) (+) T and CD(8) (+) T lymphocytes-mediated cellular immunity in pathogenesis of chronic obstructive pulmonary disease. J Immunol Res. 2022;2022:1429213. doi:10.1155/2022/1429213

22. Wu JH, Zhou M, Jin Y, et al. Generation and immune regulation of CD4(+)CD25(-)Foxp3(+) T cells in chronic obstructive pulmonary disease. Front Immunol. 2019;10:220. doi:10.3389/fimmu.2019.00220

23. Foo SY, Zhang V, Lalwani A, et al. Regulatory T cells prevent inducible BALT formation by dampening neutrophilic inflammation. J Immunol. 2015;194(9):4567–4576. doi:10.4049/jimmunol.1400909

24. Bluestone JA, Tang Q. How do CD4+CD25+ regulatory T cells control autoimmunity? Curr Opin Immunol. 2005;17(6):638–642. doi:10.1016/j.coi.2005.09.002

25. Sallusto F, Geginat J, Lanzavecchia A. Central memory and effector memory T cell subsets: function, generation, and maintenance. Annu Rev Immunol. 2004;22(1):745–763. doi:10.1146/annurev.immunol.22.012703.104702

26. Barceló B, Pons J, Ferrer JM, Sauleda J, Fuster A, Agustí AG. Phenotypic characterisation of T-lymphocytes in COPD: abnormal CD4+CD25+ regulatory T-lymphocyte response to tobacco smoking. Eur Respir J. 2008;31(3):555–562. doi:10.1183/09031936.00010407

27. Xiong XF, Zhu M, Wu HX, Fan LL, Cheng DY. Immunophenotype in acute exacerbation of chronic obstructive pulmonary disease: a cross-sectional study. Respir Res. 2022;23(1):137. doi:10.1186/s12931-022-02058-x

28. Chiu C, Openshaw PJ. Antiviral B cell and T cell immunity in the lungs. Nat Immunol. 2015;16(1):18–26. doi:10.1038/ni.3056

29. Onodera T, Takahashi Y, Yokoi Y, et al. Memory B cells in the lung participate in protective humoral immune responses to pulmonary influenza virus reinfection. Proc Natl Acad Sci U S A. 2012;109(7):2485–2490. doi:10.1073/pnas.1115369109

30. van der Strate BW, Postma DS, Brandsma CA, et al. Cigarette smoke-induced emphysema: a role for the B cell? Am J Respir Crit Care Med. 2006;173(7):751–758. doi:10.1164/rccm.200504-594OC

31. Cosio MG, Saetta M, Agusti A. Immunologic aspects of chronic obstructive pulmonary disease. N Engl J Med. 2009;360(23):2445–2454. doi:10.1056/NEJMra0804752

32. Sullivan JL, Bagevalu B, Glass C, et al. B cell-adaptive immune profile in emphysema-predominant chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2019;200(11):1434–1439. doi:10.1164/rccm.201903-0632LE

33. Seys LJ, Verhamme FM, Schinwald A, et al. Role of B cell-activating factor in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2015;192(6):706–718. doi:10.1164/rccm.201501-0103OC

34. Ladjemi MZ, Martin C, Lecocq M, et al. Increased IgA expression in lung lymphoid follicles in severe chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2019;199(5):592–602. doi:10.1164/rccm.201802-0352OC

35. Tan SY, Krasnow MA. Developmental origin of lung macrophage diversity. Development. 2016;143(8):1318–1327. doi:10.1242/dev.129122

36. Desch AN, Gibbings SL, Goyal R, et al. Flow cytometric analysis of mononuclear phagocytes in nondiseased human lung and lung-draining lymph nodes. Am J Respir Crit Care Med. 2016;193(6):614–626. doi:10.1164/rccm.201507-1376OC

37. Kapellos TS, Bassler K, Aschenbrenner AC, Fujii W, Schultze JL. Dysregulated functions of lung macrophage populations in COPD. J Immunol Res. 2018;2018:2349045. doi:10.1155/2018/2349045

38. Cros J, Cagnard N, Woollard K, et al. Human CD14dim monocytes patrol and sense nucleic acids and viruses via TLR7 and TLR8 receptors. Immunity. 2010;33(3):375–386. doi:10.1016/j.immuni.2010.08.012

39. Belge KU, Dayyani F, Horelt A, et al. The proinflammatory CD14+CD16+DR++ monocytes are a major source of TNF. J Immunol. 2002;168(7):3536–3542. doi:10.4049/jimmunol.168.7.3536

40. Russell RE, Thorley A, Culpitt SV, et al. Alveolar macrophage-mediated elastolysis: roles of matrix metalloproteinases, cysteine, and serine proteases. Am J Physiol Lung Cell Mol Physiol. 2002;283(4):L867–73. doi:10.1152/ajplung.00020.2002

41. Gnemmi I, Dossena F, Dossena F, et al.; A DIS. Pathogenesis of COPD at the cellular and molecular level. Minerva Med. 2022;113(3):405–423. doi:10.23736/s0026-4806.22.07927-7.

Creative Commons License © 2024 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.